Cyclase inhibiting parathyroid hormone antagonists or modulators and osteoporosis

The present invention relates to a novel method for treating a patient that has osteoporosis and the patient may be having administered cyclase activating parathyroid hormone (CAP) or analogues. The patient receives an administration of a cyclase inhibiting parathyroid hormone peptide (CIP) having an amino acid sequence from between (SEQ ID NO:1 [PTH2-84]) and (SEQ ID NO:3 [PTH34-84]) (i.e., a contiguous portion of PTH having an amino acid sequence set forth in SEQ ID NO:5 (PTH1-84), having the N-terminal amino acid residue starting at any position spanning from position 2 through position 34 of the PTH1-84, and the C-terminal amino acid residue ending at position 84 of the PTH1-84), (preferably (SEQ ID NO:2 [PTH3-84]) and (SEQ ID NO:8 [PTH28-84])), or a conservatively substituted variant thereof exhibiting parathyroid hormone (PTH) antagonist activity in a therapeutically effective, but non-toxic amount that reduces the occurrence of hypercalcemia or osteosarcoma in the patient resulting from the administration of CAP, and yet, through a CAP rebound effect, is effective in itself in the treatment of osteoporosis.

Skip to: Description  ·  Claims  ·  References Cited  · Patent History  ·  Patent History
Description

This application claims priority to U.S. patent application Ser. No. 60/224,446, filed Aug. 10, 2000, under 35 U.S.C. § 119(e).

TECHNICAL FIELD

The present invention relates to a novel method for treating a patient that has osteoporosis and the patient may be having administered cyclase activating parathyroid hormone (CAP) or analogues. The patient receives an administration of a cyclase inhibiting parathyroid hormone peptide (CIP) having an amino acid sequence from between (SEQ ID NO:1 [PTH2-84]) and (SEQ ID NO:3[PTH34-84]) (i.e., a contiguous portion of PTH having an amino acid sequence set forth in SEQ ID NO:5 (PTH1-84) having the N-terminal amino acid residue starting at any position spanning from position 2 through position 34 of the PTH1-84, and the C-terminal amino acid residue ending at position 84 of the PTH1-84), (preferably (SEQ ID NO:2 [PTH3-84]) and (SEQ ID NO:8 [PTH28-84])), or a conservatively substituted variant thereof exhibiting parathyroid hormone (PTH) antagonist activity in a therapeutically effective, but non-toxic amount that reduces the occurrence of hypercalcemia or osteosarcoma in the patient resulting from the administration of CAP, and yet, through a CAP rebound effect, is effective in itself in the treatment of osteoporosis.

BACKGROUND ART

Calcium plays an indispensable role in cell permeability, the formation of bones and teeth, blood coagulation, transmission of nerve impulse, and normal muscle contraction. The concentration of calcium ions in the blood is, along with calcitriol and calcitonin, regulated mainly by parathyroid hormone (PTH). Extracellular calcium levels are directly affected by PTH through calcium uptake in kidney tubule cells and calcium transport to or from bone. Although calcium intake and excretion may vary, PTH serves through a feedback mechanism to maintain a steady concentration of calcium in cells and surrounding fluids. When serum calcium lowers, the parathyroid glands secrete PTH, affecting the release of stored calcium. When serum calcium increases, stored calcium release is retarded through lowered secretions of PTH.

The complete or whole form of human PTH, (hPTH), is a unique 84 amino acid peptide (SEQ ID NO:5), as is shown in FIG. 1. Researchers have found that this peptide has an anabolic effect on bone that involves a domain for protein kinase C activation (amino acid residues 28 to 34) as well as a domain for adenylate cyclase activation (amino acid residues 1 to 7). However, various catabolic forms of clipped or fragmented PTH peptides also are found in circulation, most likely formed by intraglandular or peripheral metabolism, or example, hPTH can be cleaved between amino acids 34 and 35 to produce a (1-34) PTH N-terminal fragment and a (35-84) PTH C-terminal fragment. Likewise, clipping can occur between either amino acids 36 and 37 or 37 and 38. Recently, a large PTH fragment referred to as “non-(1-84) PTH” has been disclosed which is clipped closer to the N-terminal end of PTH. (See R. LePage et alia, “A non-(1-84) circulating parathyroid hormone (PTH) fragment interferes significantly with intact PTH commercial assay measurements in uremic samples ” Clin Chem (1998); 44: 805-810.).

The cleaved fragments of PTH vary in both biological activity and metabolic clearance rate from the circulation. For example, the N-terminal human PTH1-34 (hPTH1-34) fragment has PTH agonist properties, but is rapidly removed from circulation. A daily subcutaneous administration of hPTH to patients with idiopathic osteoporosis has been shown to substantially increase their iliac trebecular bone volume. (See R. Podbesek et alia, “Effects of two treatment regimes with synthetic human parathyroid hormone fragment on bone formation and the issue balance of trabecular bone in greyhounds”. Endocrinology (1983); 112: 1000-1006.)

Osteoporosis is the most common form of metabolic bone disease and may be considered the symptomatic, fracture stage of bone loss (osteopenia). Although osteoporosis may occur secondary to a number of underlying diseases, 90% of all cases appear to be idiopathic. Postmenopausal women are particularly at risk for idiopathic osteoporosis (postmenopausal or Type I osteoporosis). Another high risk group for idiopathic osteoporosis is the elderly of either sex (senile or Type II osteoporosis). Osteoporosis has also been related to corticosteroid use, immobilization or extended bed rest, alcoholism, diabetes, gonadotoxic chemotherapy, hyperprolactinemia, anorexia nervosa, primary and secondary amenorrhea, and oophorectomy.

In the various forms of osteoporosis, mechanical failure bone fractures frequently occur which are the result of bone loss. Postmenopausal osteoporosis is characterized by fractures of the wrist and spine, while femoral neck fractures seem to be the dominant feature of senile osteoporosis.

Bone loss in osteoporotics is believed to involve an imbalance in the process by which the skeleton renews itself This process has been termed bone remodeling. It occurs in a series of discrete pockets of activity. These pockets appear spontaneously within the bone matrix on a given bone surface as a site of bone resorption. Osteoclasts (bone dissolving or resorbing cells) are responsible for the resorption of a portion of bone of generally constant dimension. Resorption is followed by the appearance of osteoblasts (bone forming cells) that refill the cavity left by the osteoclasts with new bone.

In a healthy adult subject, the rate at which osteoclasts and osteoblasts are formed is such that bone formation and bone resorption are in balance constituting an optimal bone turnover rate. However, in osteoporotics an imbalance in the bone remodeling process develops which results in bone being lost at a rate faster than it is being made. Although this imbalance occurs to some extent in most individuals as they age, it is much more severe and occurs at a younger age in postmenopausal osteoporotics or following oophorectomy.

There have been many attempts to treat osteoporosis with the goal of either slowing further bone loss or, more desirably, producing a net gain in bone mass. Certain agents, such as estrogen and the bisphosphonates, appear to slow further bone loss in osteoporotics. Agents which slow bone loss, because of the different durations of bone resorption and formation, may appear to increase bone mass (on the order of 3% to 7%). However, this apparent increase is limited in time, not progressive, and is due to a decrease in “remodeling space.” In addition, because of the close coupling between resorption and formation, impeding bone resorption also ultimately impedes bone formation.

Another class of agents investigated to combat the onset of osteoporosis encompasses PTH and PTH analogues. (See U.S. Pat. No. 6,051,686 to Kretenansky et alia, for example.) The theory behind the use of such compounds is to use the body's natural protein receptor binding process to counter a greater removal of calcium from bone than resorption of calcium. Unfortunately, such proposed treatments have had adverse effects, including hypercalcemia (elevated serum calcium) and the formation of osteosarcomas.

DISCLOSURE OF THE INVENTION

The present invention relates to a novel method for treating a patient that has osteoporosis. The patient may be having administered cyclase activating parathyroid hormone (CAP), commonly referred to simply as PTH, or CAP analogues. The patient receives administration of a cyclase inhibiting parathyroid hormone peptide (CIP) having an amino acid sequence from between (SEQ ID NO:1 [PTH2-84]) and (SEQ ID NO:3 [PTH38-84]) (i.e., a contiguous portion of PTH having an amino acid sequence set forth in SEQ ID NO:5 (PTH1-84). having the N-terminal amino acid residue starting at any position spanning from position 2 through position 34 of the PTH1-84, and the C-terminal amino acid residue ending at position 84 of the PTH1-84), (preferably (SEQ ID NO:2 [PTH3-84]) and (SEQ ID NO:8 [PTH28-84])), or a conservatively substituted variant thereof exhibiting parathyroid hormone (PTH) antagonist activity in a therapeutically effective, but non-toxic amount that reduces the occurrence of hypercalcemia or osteosarcoma in the patient resulting from the administration of CAP. CIP also has the ability when administered alone to provide a therapeutic treatment for osteoporosis by means of the CAP rebound effect without hypercalcemia or osteosarcoma side effects. Administration can be either continuous or pulsatile, as in the administration of CAP.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 is a diagrammatic view of hPTH (SEQ ID NO:5).

FIG. 2 is a graph showing the change in serum calcium using PTH alone, the present PTH antagonist alone, a combination of PTH and present PTH antagonist, and a control.

BEST MODES FOR CARRYING OUT THE INVENTION

In disclosing the present invention, one should remember that there are a number of closely analogous, species dependent forms of PTH. The amino acid sequence of hPTH or CAP is shown in FIG. 1. However, for rat PTH, bovine PTH, or porcine PTH, for example, one finds the substitutions at some of the amino acids in the hPTH sequence. For the purposes of the present invention, one can use interchangeably truncated forms of these PTH's, although it is preferred to use a PTH having a sequence matching the species in which the PTH antagonist is used.

Preferred PTH antagonists of the present invention have an amino acid sequence from between PTH2-84 (SEQ ID NO:1) and PTH34-84 (SEQ ID NO:3) or a conservatively substituted variant thereof exhibiting PTH antagonist activity, with the most preferred form being from between PTH3-84 (SEQ ID NO:2) and PTH28-84 (SEQ ID NO:8).

PTH Antagonist Peptide Preparation

In order to make the present compositions, one can use any conventionally known method. For example, one can use recombinant DNA methods produce the desired compound.

Alternatively, one can use an automated peptide synthesizer, such as Model 431 made by Applied Biosystems, Inc. (Foster City, Calif., U.S.A.) Fmoc (9-fluoronylmethoxycarbonyl) can be used as the alpha-amino protecting group. All amino acids and solvents are available from Applied Biosystems and are of synthesis grade. Following synthesis, the peptide is cleaved from the resin, and side chains are de-blocked, using a cleavage cocktail containing 6.67% phenol, 4.4% (v/v) thioanisole and 8.8% ethanedithiol in trifluoroacetic acid (TFA). The cleaved peptide is precipitated and washed several times in cold diethyl ether. It is then dissolved in water and lyophilized. The crude peptide is subjected to amino acid analysis (Waters PICO-TAG System, Boston, Mass., U.S.A.) and reversed-phase HPLC using a VYDAC (TM) C8 column with 0.1% TFA in water and 99.9% acetonitrile in 0.1% TFA as the mobile buffers. The presence of a single major peak along with the appropriate amino acid composition is taken as evidence that the peptide is suitable for further use.

PTH Pharmaceutical Compositions

The present PTH antagonist peptides exhibit both oral and parenteral activity and can be formulated in solid or liquid dosage forms for oral, parenteral, intranasal, topical, or injectable administration using known carriers, excipients, or the like. The exact amount of present PTH antagonist used can vary depending upon the degree of antagonist property desired, the route of administration, or the duration of the treatment, as is known to the art.

Antagonist Properties

The present PTH antagonists have the ability to reduce the increase in serum calcium normally caused by PTH or a PTH agonist analog. These antagonists also possess an ability similar to CAP to treat osteoporosis due to the CAP rebound effect that comes with CIP administration. The CAP rebound effect is believed to be the body's response to an administration of CIP. This response occurs when the parathyroid gland cells secrete CAP in an effort to return the CAP/CIP ratio to homeostasis with the pre-CIP administration levels. PTH antagonist administration is not accompanied by hypercalcemia and osteosarcoma, as with direct CAP administration. FIG. 2 is a graph demonstrating such a hypercalcemic prevention property. Twenty-five rats were used in a demonstration of the effect of the present PTH antagonists. All of the rats had their parathyroid glands removed. Five rats received an i.v. injection of a saline control. The serum calcium of the control rats was measured and on average was lowered over time by about 0.18 mg/dl by virtue of a parathyroidectomy. Nine rats received an i.v. injection (10 μg/kg) of hPTH obtained from Bachem, A G of Bubendorf, Switzerland. The serum calcium of the hPTH rats was measured and on average was raised over time by about 0.65 mg/dl. Five rats received an equimolar i.v. injection of a PTH7-84 (the PTH antagonist) also obtained from from Bachem, A G of Bubendorf, Switzerland. The serum calcium of the PTH antagonist rats was measured and on average was lowered over time by about 0.30 mg/dl. Finally, six rats received an i.v. injection comprised of hPTH (10 μg/kg) and an equimolar amount of the PTH antagonist PTH7-84. The serum calcium of the hPTH/PTH antagonist rats was measured and on average remained substantially the same over time, raising only about 0.03 mg/dl. Thus, the present composition was able to prevent the substantial serum calcium increase normally associated with an administration of hPTH to rats having hypoparathyroidism, and quite unexpectedly, is much more potent in its antagonist property than the previously reported antagonist PTH3-34.

The ordinarily skilled artisan can appreciate that the present invention can incorporate any number of the preferred features described above.

All publications or unpublished patent applications mentioned herein are hereby incorporated by reference thereto.

Other embodiments of the present invention are not presented here which are obvious to those of ordinary skill in the art, now or during the term of any patent issuing from this patent specification, and thus, are within the spirit and scope of the present invention.

Claims

1. A method for reducing the occurrence of hypercalcemia or osteosarcoma in a patient,

wherein said patient has osteoporosis and has received administration of, or is being administered, cyclase activating parathyroid hormone (CAP) or analogues thereof,
said method comprising also administering a cyclase inhibiting parathyroid hormone peptide (CIP) to said patient in a therapeutically effective, but non-toxic amount,
where said CIP comprises a contiguous portion of PTH having amino acid sequence set forth in SEQ ID NO:5 (PTH1-84), said portion having an N-terminal amino acid residue starting at any position spanning from position 2 through position 34 of the PTH1-84, and a C-terminal amino acid residue ending at position 84 of the PTH1-84, or a conservatively substituted variant thereof exhibiting parathyroid hormone (PTH) antagonist activity.

2. The method of claim 1 wherein the CIP has an N-terminal amino acid residue starting at any position spanning from position 3 through position 28 of the PTH1-84, and a C-terminal amino acid residue ending at position 84 of the PTH1-84.

3. The method of claim 1, wherein the CIP administered is PTH7-84.

4. The method of claim 1 wherein the CIP administration is performed in a pulsatile manner.

5. The method of claim 1, wherein the CIP is administered to reduce hypercalcemia.

6. The method of claim 1, wherein the CIP is administered to reduce osteosarcomas.

7. A method for reducing the occurrence of hypercalcemia or osteosarcoma in a patient,

wherein said patient has osteoporosis and has received administration of, or is being administered, cyclase activating parathyroid hormone (CAP) or analogues thereof,
said method comprising also administering a cyclase inhibiting parathyroid hormone peptide (CIP) to said patient in a therapeutically effective, but non-toxic amount,
where said CIP comprises a contiguous portion of PTH having an amino acid sequence set forth in SEQ ID NO:5 (PTH1-84), said portion having an N-terminal amino acid residue starting at any position spanning from position 2 through position 34 of the PTH1-84, and a C-terminal amino acid residue ending at position 84 of the PTH1-84.
Referenced Cited
U.S. Patent Documents
4369138 January 18, 1983 Lindall
4423037 December 27, 1983 Rosenblatt et al.
4508828 April 2, 1985 Lindall et al.
4656250 April 7, 1987 Morita et al.
5856138 January 5, 1999 Fukuda
6030790 February 29, 2000 Adermann et al.
6051686 April 18, 2000 Krstenansky et al.
Foreign Patent Documents
33 47 548 July 1985 DE
44 34 551 April 1996 DE
0 783 522 December 2001 EP
WO 91 06564 May 1991 WO
EP 0 451 867 November 1991 WO
WO 93 06845 April 1993 WO
WO 94 03201 February 1994 WO
Other references
  • Chorey et al., Biochemistry (1990) 29:1580-1586.
  • Hoare et al., Journal of Biological Chemistry (2000) 275(35):27274-27283.
  • International Search Report for PCT/US02/25348, mailed on Jul. 26, 2004, 4 pages.
  • Jonsson et al., Endocrinology (2001) 142(2):704-709.
  • Pausova et al., Mammalian Genome (1995) 6:408-414.
  • Takasu et al., Journal of Bone Mineral Metabolism (1994) 12(Suppl.1):S131-S134.
  • Yu et al., Endocrinology (1997) 138(8):3085-3092.
  • U.S. Appl. No. 09/231/422, filed Jan. 14, 1999.
  • U.S. Appl. No. 09/344,639, filed Jun. 26, 1999.
  • U.S. Appl. No. 09/928,047, filed Aug. 10, 2001.
  • U.S. Appl. No. 09/636,530, filed Aug. 10, 2001.
  • U.S. Appl. No. 09/636,531, filed Aug. 10, 2000.
  • U.S. Appl. No. 10/002,818, filed Nov. 2, 2001.
  • U.S. Appl. No. 60/224,396, filed Aug. 10, 2000.
  • U.S. Appl. No. 09/928,048, filed Aug. 10, 2001.
  • U.S. Appl. No. 09/323,606, filed Jun. 2, 1999.
  • Aldermann et al., in: Innovations and Perspectives in Solid Phase Synthesis, Epton (ed.). Mayflower World Wide, Birmingham (1994) pp. 429-432.
  • Atkinson et al., Journal of Immunoassay (1982) 3(1):31-51.
  • Blind et al., Clin. Chem. (1987) 33(8):1376-1381.
  • Bowie et al., Science (1990) 247:1306-1310.
  • Brossard et al., Journal of Clinical Endocrinology and Metabolism (1996) 81(11):3923-3929.
  • Campbell, Monoclonal Antibody and Immunosensor Technology, in Laboratory Techniques in Biochemistry and Molecular Biology, van der Vliet (ed.), Elsevier (1991) pp. 1-11, 42-45.
  • Caporale and Rosenblatt, Paraththyroid Hormone Antagonists Effective in vivo, in: Advances in Experimental Medicine and Biology, New York (1986) pp. 315-327.
  • Clinical Chemistry (1999) 45(6)Suppl:A97 b. Abstract Nos. 339-341.
  • D'Amour et al., Am. J. Physiol. (1986) 251:E680-E687.
  • Daniel et al., Virology (1994) 202:540-549.
  • Divieti, P. et al. (2001). J Bone Miner Res 2001:Suppl 1, S307.
  • Faugere, M.C. et al. (2001). Kidney International 60:1-460-1-468.
  • Faugere, M.C. et al. Nephrology, Bone & Mineral Metabolism A3995.
  • Fischer et al., The Journal of Clinical Investigation (1974) 54:1382-1394.
  • Gao et al., Clinica Chimica Acta (1996) 245:39-59.
  • Goodman, W. et al. (2000). NEJM 342:20, 1478-1483.
  • Gordon et al., Parathyroid Hormone Domain for Protein Kinase C Stimulation Located within Amphiphilic Helix, in: Peptides: Chemistry and Biology, Proceedings of the Twelfth American Peptide Symposium, Jun. 16-21, 1991. Cambridge, MA, Smith and Rivier (eds.) Escom Science Publishers (1992) pp. 37-39.
  • Hashimoto et al., Journal of Cardiovascular Pharmacology (1981) 3(4):668-676.
  • Hehrmann et al., Journal of Immunoassay (1980) 1(2):151-174.
  • John et al., Journal of Clinical Endocrinology and Metabolism (1999) 84(11):4287-4290.
  • LePage et al., Clin. Chem. (1998) 44:805-810.
  • Logue et al., Journal of Immunological Methods (1991) 137:159-166.
  • Mägerlein et al., Arzneim.-Forsch. Drug Res. (1998) 48(1):197-204.
  • Mägerlein et al., Arzneim.-Forsch. Drug Res. (1998) 48(11):783-787.
  • Mallette, Journal of Clinical Endocrinology and Metabolism (1980) 50(1):201-203.
  • Nakamura et al., Endocrinol. JPN (1981) 28(4):547-549.
  • Ngo et al., The Protein Folding Problem and Tertiary Structure Prediction. Merz et al., (eds.). Birkhäuser Boston (1994) pp. 492-495.
  • Niall et al., Proc. Natl. Acad. Sci. USA (1974) 71(2):384-388.
  • Nussbaum et al., Chemical Abstracts (1982) 96(5):181-192.
  • Pang et al., Pharmacol. Exp. Ther. (1981) 216(3):567-571.
  • Podbesek et al. (1983). Endocrinology 112(3):1000-1006.
  • Qi et al., Am. J. Kidney Dis. (1995) 26:622-631.
  • Quarles et al., J. Clin. Endocrinol. Metab. (1992) 75:145-150.
  • Stadler, Homologous Radioimmunoassay for Human Parathyroid Hormone (Residues 1-34) with Biotinylated Peptide as Tracer, in Calcium Regulating Hormones, Vitamin D Metabolites, and Cyclic AMP Assays and their Clinical Application, Schmidt-Gayk et al., (eds.). Berlin/Heidelberg, Springer, (1990) pp. 137-150.
  • Tampe et al., J. Immunoassay (1992) 13(1):1-13.
  • Visser et al., Acta Endocrinology (1979) 90:90-102.
  • Wingender et al., Structure-Function Relationship in Parathyroid Hormone in: Advances in Protein Design, International Workshop, Blöcker et al. (eds.). VCH (1988) pp. 167-176.
  • Zanelli et al., Journal of Immunoassay (1983) 4(2):175-206.
  • Zemplar package insert. Abbott Reference (1998), 06-9998-R1-Rev. Roche Laboratories.
Patent History
Patent number: 6923968
Type: Grant
Filed: Aug 10, 2001
Date of Patent: Aug 2, 2005
Patent Publication Number: 20020160945
Assignee: Scantibodies Laboratory, Inc. (Santee, CA)
Inventor: Thomas L. Cantor (El Cajon, CA)
Primary Examiner: Lorraine Spector
Assistant Examiner: Dong Jiang
Attorney: Morrison & Foerster LLP
Application Number: 09/928,047